1
|
Six KR, Vertongen S, Seghers S, De Bleser D, Compernolle V, Feys HB. Differential composition and yield of leukocytes isolated from various blood component leukoreduction filters. J Immunol Methods 2024; 533:113733. [PMID: 39098592 DOI: 10.1016/j.jim.2024.113733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 08/06/2024]
Abstract
In Flanders, an estimated 300,000 leukoreduction filters are discarded as biological waste in the blood establishment each year. These filters are a possible source of fresh donor leukocytes for downstream purposes including research. We investigated leukocyte isolation from two types of filters either used for the preparation of platelet concentrates (PC-LRF) or erythrocyte concentrates (EC-LRF). Outcome parameters were leukocyte yield, differential count, turnaround time and effect of storage conditions. Leukocytes were harvested by reverse flow of a buffer solution. Control was the gold standard density gradient centrifugation of buffy coats. Total leukocyte number isolated from PC-LRF (1049 (± 40) x 106) was almost double that of control (632 (± 66) x 106) but the differential count was comparable. Total leukocyte number isolated from EC-LRF (78 (± 9) x 106) was significantly lower than control, but the sample was specifically enriched in granulocytes (81 ± 4%) compared to control (30 ± 1%). Isolation of leukocytes from either PC- or EC-LRF takes 20 min compared to 240 min for control density gradient centrifugation. Leukocyte viability is optimal when harvested on day 1 post donation (95 ± 0.9%) compared to day 3 (76.4 ± 2.4%). In conclusion, our study demonstrates that leukoreduction filters from specific blood component processing are easy to use and present a valuable source for viable leukocytes of all types.
Collapse
Affiliation(s)
- Katrijn R Six
- Transfusion Research Center, Belgian Red Cross Flanders, Ghent, Belgium.
| | - Sarah Vertongen
- Transfusion Research Center, Belgian Red Cross Flanders, Ghent, Belgium
| | - Sabrina Seghers
- Transfusion Innovation Center, Belgian Red Cross Flanders, Ghent, Belgium
| | | | - Veerle Compernolle
- Transfusion Research Center, Belgian Red Cross Flanders, Ghent, Belgium; Transfusion Innovation Center, Belgian Red Cross Flanders, Ghent, Belgium; Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium; Blood Services, Belgian Red Cross Flanders, Mechelen, Belgium
| | - Hendrik B Feys
- Transfusion Research Center, Belgian Red Cross Flanders, Ghent, Belgium; Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
2
|
Chen S, Nguyen TD, Lee KZ, Liu D. Ex vivo T cell differentiation in adoptive immunotherapy manufacturing: Critical process parameters and analytical technologies. Biotechnol Adv 2024; 77:108434. [PMID: 39168355 DOI: 10.1016/j.biotechadv.2024.108434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 08/01/2024] [Accepted: 08/17/2024] [Indexed: 08/23/2024]
Abstract
Adoptive immunotherapy shows great promise as a treatment for cancer and other diseases. Recent evidence suggests that the therapeutic efficacy of these cell-based therapies can be enhanced by the enrichment of less-differentiated T cell subpopulations in the therapeutic product, giving rise to a need for advanced manufacturing technologies capable of enriching these subpopulations through regulation of T cell differentiation. Studies have shown that modifying certain critical process control parameters, such as cytokines, metabolites, amino acids, and culture environment, can effectively manipulate T cell differentiation in ex vivo cultures. Advanced process analytical technologies (PATs) are crucial for monitoring these parameters and the assessment of T cell differentiation during culture. In this review, we examine such critical process parameters and PATs, with an emphasis on their impact on enriching less-differentiated T cell population. We also discuss the limitations of current technologies and advocate for further efforts from the community to establish more stringent critical process parameters (CPPs) and develop more at-line/online PATs that are specific to T cell differentiation. These advancements will be essential to enable the manufacturing of more efficacious adoptive immunotherapy products.
Collapse
Affiliation(s)
- Sixun Chen
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, 20 Biopolis Way, 138668, Singapore
| | - Tan Dai Nguyen
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, 20 Biopolis Way, 138668, Singapore
| | - Kang-Zheng Lee
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, 20 Biopolis Way, 138668, Singapore
| | - Dan Liu
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, 20 Biopolis Way, 138668, Singapore.
| |
Collapse
|
3
|
Hood T, Slingsby F, Sandner V, Geis W, Schmidberger T, Bevan N, Vicard Q, Hengst J, Springuel P, Dianat N, Rafiq QA. A quality-by-design approach to improve process understanding and optimise the production and quality of CAR-T cells in automated stirred-tank bioreactors. Front Immunol 2024; 15:1335932. [PMID: 38655265 PMCID: PMC11035805 DOI: 10.3389/fimmu.2024.1335932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
Ex vivo genetically-modified cellular immunotherapies, such as chimeric antigen receptor T cell (CAR-T) therapies, have generated significant clinical and commercial outcomes due to their unparalleled response rates against relapsed and refractory blood cancers. However, the development and scalable manufacture of these novel therapies remains challenging and further process understanding and optimisation is required to improve product quality and yield. In this study, we employ a quality-by-design (QbD) approach to systematically investigate the impact of critical process parameters (CPPs) during the expansion step on the critical quality attributes (CQAs) of CAR-T cells. Utilising the design of experiments (DOE) methodology, we investigated the impact of multiple CPPs, such as number of activations, culture seeding density, seed train time, and IL-2 concentration, on CAR-T CQAs including, cell yield, viability, metabolism, immunophenotype, T cell differentiation, exhaustion and CAR expression. Initial studies undertaken in G-Rex® 24 multi-well plates demonstrated that the combination of a single activation step and a shorter, 3-day, seed train resulted in significant CAR-T yield and quality improvements, specifically a 3-fold increase in cell yield, a 30% reduction in exhaustion marker expression and more efficient metabolism when compared to a process involving 2 activation steps and a 7-day seed train. Similar findings were observed when the CPPs identified in the G-Rex® multi-well plates studies were translated to a larger-scale automated, controlled stirred-tank bioreactor (Ambr® 250 High Throughput) process. The single activation step and reduced seed train time resulted in a similar, significant improvement in CAR-T CQAs including cell yield, quality and metabolism in the Ambr® 250 High Throughput bioreactor, thereby validating the findings of the small-scale studies and resulting in significant process understanding and improvements. This study provides a methodology for the systematic investigation of CAR-T CPPs and the findings demonstrate the scope and impact of enhanced process understanding for improved CAR-T production.
Collapse
Affiliation(s)
- Tiffany Hood
- Department of Biochemical Engineering, University College London, London, United Kingdom
| | - Fern Slingsby
- Product Excellence Bioreactor Technology, Sartorius Stedim UK Limited, Epsom, United Kingdom
| | - Viktor Sandner
- Digital Solutions, Sartorius Stedim Austria GmbH, Vienna, Austria
| | - Winfried Geis
- Digital Solutions, Sartorius Stedim Biotech GmbH, Goettingen, Germany
| | - Timo Schmidberger
- Digital Solutions, Sartorius Stedim Biotech GmbH, Goettingen, Germany
| | - Nicola Bevan
- BioAnalytics Application Development, Essen BioScience Ltd. (Part of the Sartorius Group), Royston, United Kingdom
| | - Quentin Vicard
- Cell Culture Technology Marketing, Sartorius Stedim France S.A.S., Aubagne, France
| | - Julia Hengst
- Cell Culture Technology Marketing, Sartorius Stedim Biotech GmbH, Goettingen, Germany
| | - Pierre Springuel
- Department of Biochemical Engineering, University College London, London, United Kingdom
| | - Noushin Dianat
- Cell Culture Technology Marketing, Sartorius Stedim France S.A.S., Aubagne, France
| | - Qasim A. Rafiq
- Department of Biochemical Engineering, University College London, London, United Kingdom
| |
Collapse
|
4
|
Poiret T, Vikberg S, Schoutrop E, Mattsson J, Magalhaes I. CAR T cells and T cells phenotype and function are impacted by glucocorticoid exposure with different magnitude. J Transl Med 2024; 22:273. [PMID: 38475830 PMCID: PMC10935894 DOI: 10.1186/s12967-024-05063-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 03/05/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T cell therapy is associated with high risk of adverse events. Glucocorticoids (GCs) are cornerstone in the management of high-grade cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Given the potentially deleterious effects of GCs on CAR T cells anti-tumor activity, increasing our understanding of GCs impact on CAR T cells is crucial. METHODS Using several CAR T cells i.e., CD19, mesothelin (MSLN)-CD28 and MSLN-41BB CAR T cells (M28z and MBBz), we compared phenotypical, functional, changes and anti-tumor activity between i) transduced CD19 CAR T cells with untransduced T cells, ii) M28z with MBBz CAR T cells induced by Dexamethasone (Dx) or Methylprednisolone (MP) exposures. RESULTS Higher levels of GC receptor were found in less differentiated CAR T cells. Overall, Dx and MP showed a similar impact on CAR T cells. Compared to untreated condition, GCs exposure increased the expression of PD-1 and TIM-3 and reduced the expression of LAG3 and function of T cells and CAR T cells. GC exposures induced more exhausted (LAG3 + PD1 + TIM3 +) and dysfunctional (CD107a-INFγ-TNF-IL2-) untransduced T cells in comparison to CD19 CAR T cells. GC exposure impaired more CD4 + than CD8 + CD19 CAR T cells. GC exposures increased more PD-1 expression associated with reduced proliferative capacity and function of M28z as compared to MBBz CAR T cells. CAR T cells anti-tumor activity was greatly affected by repeated GC exposure but partly recovered within 48h after GCs withdrawal. CONCLUSIONS In summary, GCs impacted phenotype and function of untransduced and CAR T cell with different magnitude. The nature of the CAR costimulatory domain influenced the magnitude of CAR T cell response to GCs.
Collapse
Affiliation(s)
- Thomas Poiret
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| | - Sara Vikberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Esther Schoutrop
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Mattsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Gloria and Seymour Epstein Chair in Cell Therapy and Transplantation, Princess Margaret Cancer Centre and University of Toronto; Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Isabelle Magalhaes
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
5
|
Sanyanusin M, Tudsamran S, Thaiwong R, Tawinwung S, Nishio N, Takahashi Y, Hirankarn N, Suppipat K. Novel xeno-free and serum-free culturing condition to improve piggyBac transposon-based CD19 chimeric antigen receptor T-cell production and characteristics. Cytotherapy 2023; 25:397-406. [PMID: 36517366 DOI: 10.1016/j.jcyt.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/22/2022] [Accepted: 11/27/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND AIMS Chimeric antigen receptor (CAR) T cell is a novel therapy for relapse and refractory hematologic malignancy. Characteristics of CAR T cells are associated with clinical efficacy and toxicity. The type of serum supplements used during cultivation affects the immunophenotype and function of viral-based CAR T cells. This study explores the effect of serum supplements on nonviral piggyBac transposon CAR T-cell production. METHODS PiggyBac CD19 CAR T cells were expanded in cultured conditions containing fetal bovine serum, human AB serum or xeno-free serum replacement. We evaluated the effect of different serum supplements on cell expansion, transduction efficiency, immunophenotypes and antitumor activity. RESULTS Xeno-free serum replacement exhibited comparable CAR surface expression, cell expansion and short-term antitumor activity compared with conventional serum supplements. However, CAR T cells cultivated with xeno-free serum replacement exhibited an increased naïve/stem cell memory population and better T-cell expansion after long-term co-culture as well as during the tumor rechallenge assay. CONCLUSIONS Our study supports the usage of xeno-free serum replacement as an alternative source of serum supplements for piggyBac-based CAR T-cell expansion.
Collapse
Affiliation(s)
- Mulita Sanyanusin
- Medical Microbiology, Interdisciplinary and International Program, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Suparat Tudsamran
- Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand; Chulalongkorn Comprehensive Cancer Center, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Rattapoom Thaiwong
- Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand; Chulalongkorn Comprehensive Cancer Center, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Supannikar Tawinwung
- Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand; Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Nobuhiro Nishio
- Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Yoshiyuki Takahashi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nattiya Hirankarn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Koramit Suppipat
- Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand; Chulalongkorn Comprehensive Cancer Center, King Chulalongkorn Memorial Hospital, Bangkok, Thailand; Department of Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
6
|
Schoutrop E, Poiret T, El-Serafi I, Zhao Y, He R, Moter A, Henriksson J, Hassan M, Magalhaes I, Mattsson J. Tuned activation of MSLN-CAR T cells induces superior antitumor responses in ovarian cancer models. J Immunother Cancer 2023; 11:jitc-2022-005691. [PMID: 36746513 PMCID: PMC9906404 DOI: 10.1136/jitc-2022-005691] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Limited persistence of functional CAR T cells in the immunosuppressive solid tumor microenvironment remains a major hurdle in the successful translation of CAR T cell therapy to treat solid tumors. Fine-tuning of CAR T cell activation by mutating CD3ζ chain immunoreceptor tyrosine-based activation motifs (ITAMs) in CD19-CAR T cells (containing the CD28 costimulatory domain) has proven to extend functional CAR T cell persistence in preclinical models of B cell malignancies. METHODS In this study, two conventional second-generation MSLN-CAR T cell constructs encoding for either a CD28 co-stimulatory (M28z) or 4-1BB costimulatory (MBBz) domain and a novel mesothelin (MSLN)-directed CAR T cell construct encoding for the CD28 costimulatory domain and CD3ζ chain containing a single ITAM (M1xx) were evaluated using in vitro and in vivo preclinical models of ovarian cancer. Two ovarian cancer cell lines and two orthotopic models of ovarian cancer in NSG mice were used: SKOV-3 cells inoculated through microsurgery in the ovary and to mimic a disseminated model of advanced ovarian cancer, OVCAR-4 cells injected intraperitoneally. MSLN-CAR T cell treatment efficacy was evaluated by survival analysis and the characterization and quantification of the different MSLN-CAR T cells were performed by flow cytometry, quantitative PCR and gene expression analysis. RESULTS M1xx CAR T cells elicited superior antitumor potency and persistence, as compared with the conventional second generation M28z and MBBz CAR T cells. Ex vivo M28z and MBBz CAR T cells displayed a more exhausted phenotype than M1xx CAR T cells as determined by co-expression of PD-1, LAG-3 and TIM-3. Furthermore, M1xx CAR T cells showed superior ex vivo IFNy, TNF and GzB production and were characterized by a self-renewal gene signature. CONCLUSIONS Altogether, our study demonstrates the enhanced therapeutic potential of MSLN-CAR T cells expressing a mutated CD3ζ chain containing a single ITAM for the treatment of ovarian cancer. CAR T cells armored with calibrated activation potential may improve the clinical responses in solid tumors.
Collapse
Affiliation(s)
- Esther Schoutrop
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Poiret
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Ibrahim El-Serafi
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden,Basic Medical Sciences Department, College of Medicine, Ajman University, Ajman, UAE,Department of Biochemistry, Faculty of Medicine, Port-Said University, Port-Said, Egypt
| | - Ying Zhao
- Experimental Cancer Medicine, Division of Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden,Clinical Research Center and Center of Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Rui He
- Experimental Cancer Medicine, Division of Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden,Clinical Research Center and Center of Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Alina Moter
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Johan Henriksson
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Moustapha Hassan
- Experimental Cancer Medicine, Division of Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden,Clinical Research Center and Center of Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Isabelle Magalhaes
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden .,Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Mattsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden,Gloria and Seymour Epstein Chair in Cell Therapy and Transplantation, Princess Margaret Cancer Centre and University of Toronto, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Liu C, Liu AJ. Structural Characterization of an Alcohol-Soluble Polysaccharide from Bletilla striata and Antitumor Activities in Vivo and in Vitro. Chem Biodivers 2022; 19:e202200635. [PMID: 36282907 DOI: 10.1002/cbdv.202200635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/25/2022] [Indexed: 01/07/2023]
Abstract
In general, Bletilla striata polysaccharides were mostly water-soluble. However, the structural property, immunomodulatory effects and antitumor activities of alcohol-soluble Bletilla striata polysaccharide were rarely reported. In this study, an alcohol-soluble Bletilla striata polysaccharide was firstly extracted, investigated the structural property and evaluated the antitumor activity in vivo and in vitro. Results showed that BSAP was a low molecular weight polysaccharide (2.29×104 Da) and consisted of glucose, xylose and mannose (molar ratio: 2.39 : 1.00 : 0.21). Animal experiments results suggested that BSAP could effectively inhibit the expansion of H22 solid tumors, protect thymus and spleen, improve macrophages, lymphocytes and NK cells activities and enhance lymphocyte subsets proportion, presenting a better immunological enhancement effect in vivo. Additionally, the results of cell experiments showed that BSAP had obvious antitumor effect in vitro, including inhibiting the proliferation of H22 cells and inducing the apoptosis of tumor cells. These results would provide theoretical basis and new ideas for the further development and utilization of BSAP in the biomedical field.
Collapse
Affiliation(s)
- Chao Liu
- College of Food Science and Engineering, Tianjin University of Science and Technology, No. 29, 13th Street, TEDA, Tianjin, 300457, P. R. China
| | - An-Jun Liu
- College of Food Science and Engineering, Tianjin University of Science and Technology, No. 29, 13th Street, TEDA, Tianjin, 300457, P. R. China
| |
Collapse
|
8
|
Sudarsanam H, Buhmann R, Henschler R. Influence of Culture Conditions on Ex Vivo Expansion of T Lymphocytes and Their Function for Therapy: Current Insights and Open Questions. Front Bioeng Biotechnol 2022; 10:886637. [PMID: 35845425 PMCID: PMC9277485 DOI: 10.3389/fbioe.2022.886637] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/16/2022] [Indexed: 01/03/2023] Open
Abstract
Ex vivo expansion of T lymphocytes is a central process in the generation of cellular therapies targeted at tumors and other disease-relevant structures, which currently cannot be reached by established pharmaceuticals. The influence of culture conditions on T cell functions is, however, incompletely understood. In clinical applications of ex vivo expanded T cells, so far, a relatively classical standard cell culture methodology has been established. The expanded cells have been characterized in both preclinical models and clinical studies mainly using a therapeutic endpoint, for example antitumor response and cytotoxic function against cellular targets, whereas the influence of manipulations of T cells ex vivo including transduction and culture expansion has been studied to a much lesser detail, or in many contexts remains unknown. This includes the circulation behavior of expanded T cells after intravenous application, their intracellular metabolism and signal transduction, and their cytoskeletal (re)organization or their adhesion, migration, and subsequent intra-tissue differentiation. This review aims to provide an overview of established T cell expansion methodologies and address unanswered questions relating in vivo interaction of ex vivo expanded T cells for cellular therapy.
Collapse
Affiliation(s)
| | | | - Reinhard Henschler
- Institute of Transfusion Medicine, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
9
|
Schoutrop E, Renken S, Micallef Nilsson I, Hahn P, Poiret T, Kiessling R, Wickström SL, Mattsson J, Magalhaes I. Trogocytosis and fratricide killing impede MSLN-directed CAR T cell functionality. Oncoimmunology 2022; 11:2093426. [PMID: 35898704 PMCID: PMC9313125 DOI: 10.1080/2162402x.2022.2093426] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Successful translation of chimeric antigen receptor (CAR) T cell therapy for the treatment of solid tumors has proved to be troublesome, mainly due to the complex tumor microenvironment promoting T cell dysfunction and antigen heterogeneity. Mesothelin (MSLN) has emerged as an attractive target for CAR T cell therapy of several solid malignancies, including ovarian cancer. To improve clinical response rates with MSLN-CAR T cells, a better understanding of the mechanisms impacting CAR T cell functionality in vitro is crucial. Here, we demonstrated superior cytolytic capacity of CD28-costimulated MSLN-CAR T cells (M28z) relative to 4–1BB-costimulated MSLN-CAR T cells (MBBz). Furthermore, CD28-costimulated MSLN CAR T cells displayed enhanced cytolytic capacity against tumor spheroids with heterogeneous MSLN expression compared to MBBz CAR T cells. In this study, we identified CAR-mediated trogocytosis as a potential impeding factor for successful MSLN-CAR T cell therapy due to fratricide killing and contributing to tumor antigen heterogeneity. Moreover, we link antigen-dependent upregulation of LAG-3 with reduced CAR T cell functionality. Taken together, our study highlights the therapeutic potential and bottlenecks of MSLN-CAR T cells, providing a rationale for combinatorial treatment strategies.
Collapse
Affiliation(s)
- Esther Schoutrop
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Stefanie Renken
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | - Paula Hahn
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Poiret
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Rolf Kiessling
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Theme Cancer, Patient Area Head and Neck, Lung and Skin, Karolinska University Hospital, Stockholm, Sweden
| | - Stina L Wickström
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Theme Cancer, Patient Area Head and Neck, Lung and Skin, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Mattsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Gloria and Seymour Epstein Chair in Cell Therapy and Transplantation, Princess Margaret Cancer Centre and University of Toronto, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Isabelle Magalhaes
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
10
|
Watanabe N, Mo F, McKenna MK. Impact of Manufacturing Procedures on CAR T Cell Functionality. Front Immunol 2022; 13:876339. [PMID: 35493513 PMCID: PMC9043864 DOI: 10.3389/fimmu.2022.876339] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/15/2022] [Indexed: 12/21/2022] Open
Abstract
The field of chimeric antigen receptor (CAR) modified T cell therapy has rapidly expanded in the past few decades. As of today, there are six CAR T cell products that have been approved by the FDA: KYMRIAH (tisagenlecleucel, CD19 CAR T cells), YESCARTA (axicabtagene ciloleucel, CD19 CAR T cells), TECARTUS (brexucabtagene autoleucel, CD19 CAR T cells), BREYANZI (lisocabtagene maraleucel, CD19 CAR T cells), ABECMA (idecabtagene vicleucel, BCMA CAR T cells) and CARVYKTI (ciltacabtagene autoleucel, BCMA CAR T cells). With this clinical success, CAR T cell therapy has become one of the most promising treatment options to combat cancers. Current research efforts focus on further potentiating its efficacy in non-responding patients and solid tumor settings. To achieve this, recent evidence suggested that, apart from developing next-generation CAR T cells with additional genetic modifications, ex vivo culture conditions could significantly impact CAR T cell functionality - an often overlooked aspect during clinical translation. In this review, we focus on the ex vivo manufacturing process for CAR T cells and discuss how it impacts CAR T cell function.
Collapse
Affiliation(s)
- Norihiro Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Feiyan Mo
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Mary Kathryn McKenna
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
11
|
Lu Y, Zhang J, Chen Y, Kang Y, Liao Z, He Y, Zhang C. Novel Immunotherapies for Osteosarcoma. Front Oncol 2022; 12:830546. [PMID: 35433427 PMCID: PMC9012135 DOI: 10.3389/fonc.2022.830546] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/28/2022] [Indexed: 02/05/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone sarcoma mainly affecting adolescents and young adults, which often progresses to pulmonary metastasis and leads to the death of OS patients. OS is characterized as a highly heterogeneous cancer type and the underlying pathologic mechanisms triggering tumor progress and metastasis are incompletely recognized. Surgery combined with neoadjuvant and postoperative chemotherapy has elevated 5-year survival to over 70% for patients with localized OS tumors, as opposed to only 20% of patients with recurrence and/or metastasis. Therefore, novel therapeutic strategies are needed to overcome the drawbacks of conventional treatments. Immunotherapy is gaining momentum for the treatment of OS with an increasing number of FDA-approved therapies for malignancies resistant to conventional therapies. Here, we review the OS tumor microenvironment and appraise the promising immunotherapies available in the management of OS.
Collapse
Affiliation(s)
- Yubao Lu
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiahe Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yutong Chen
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yuchen Kang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Zhipeng Liao
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yuanqi He
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Cangyu Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- *Correspondence: Cangyu Zhang,
| |
Collapse
|
12
|
Schoutrop E, El-Serafi I, Poiret T, Zhao Y, Gultekin O, He R, Moyano-Galceran L, Carlson JW, Lehti K, Hassan M, Magalhaes I, Mattsson J. Mesothelin-Specific CAR T Cells Target Ovarian Cancer. Cancer Res 2021; 81:3022-3035. [PMID: 33795251 DOI: 10.1158/0008-5472.can-20-2701] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 02/02/2021] [Accepted: 03/26/2021] [Indexed: 11/16/2022]
Abstract
New therapeutic options for patients with ovarian cancer are urgently needed. Therefore, we evaluated the efficacy of two second-generation mesothelin (MSLN)-directed CAR T cells in orthotopic mouse models of ovarian cancer. Treatment with CAR T cells expressing an MSLN CAR construct including the CD28 domain (M28z) significantly prolonged survival, but no persistent tumor control was observed. Despite lower response rates, MSLN-4-1BB (MBBz) CAR T cells induced long-term remission in some SKOV3-bearing mice. Tumor-infiltrating M28z and MBBz CAR T cells upregulated PD-1 and LAG3 in an antigen-dependent manner while MSLN+ tumor cells expressed the corresponding ligands (PD-L1 and HLA-DR), demonstrating that coinhibitory pathways impede CAR T-cell persistence in the ovarian tumor microenvironment. Furthermore, profiling plasma soluble factors identified a cluster of M28z- and MBBz-treated mice characterized by elevated T-cell secreted factors that had increased survival, higher CD8+ T-cell tumor infiltration, less exhausted CAR T-cell phenotypes, and increased HLA-DR expression by tumor cells. Altogether, our study demonstrates the therapeutic potential of MSLN-CAR T cells to treat ovarian cancer. SIGNIFICANCE: These findings demonstrate that MSLN-directed CAR T cells can provide antitumor immunity against ovarian cancer.
Collapse
Affiliation(s)
- Esther Schoutrop
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Ibrahim El-Serafi
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Biochemistry, Faculty of Medicine, Port-Said University, Port-Said, Egypt
| | - Thomas Poiret
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Ying Zhao
- Experimental Cancer Medicine, Division of Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Research Center and Center of Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Okan Gultekin
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Rui He
- Experimental Cancer Medicine, Division of Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Research Center and Center of Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Lidia Moyano-Galceran
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Joseph W Carlson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Kaisa Lehti
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Biomedical Laboratory Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Moustapha Hassan
- Experimental Cancer Medicine, Division of Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Research Center and Center of Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Isabelle Magalhaes
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
- Department of Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Mattsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
- Gloria and Seymour Epstein Chair in Cell Therapy and Transplantation, Princess Margaret Cancer Centre and University of Toronto; Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
13
|
Simoni Y, Becht E, Li S, Loh CY, Yeong JPS, Lim TKH, Takano A, Tan DSW, Newell EW. Partial absence of PD-1 expression by tumor-infiltrating EBV-specific CD8 + T cells in EBV-driven lymphoepithelioma-like carcinoma. Clin Transl Immunology 2020; 9:e1175. [PMID: 32995000 PMCID: PMC7503213 DOI: 10.1002/cti2.1175] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 07/15/2020] [Accepted: 08/10/2020] [Indexed: 12/11/2022] Open
Abstract
Objectives Lymphoepithelioma‐like carcinoma (LELC) is an uncommon lung cancer, typically observed in young, non‐smoking Asian populations. LELC is associated with Epstein–Barr virus (EBV) infection of lung tumor cells of epithelial origin, suggesting a carcinogenic role of EBV as observed in nasopharyngeal carcinoma (NPC). Here, we studied the antigen specificity and phenotype of EBV‐specific CD8+ T cells in blood and tumor of one LELC patient positive for EBV infection in lung tumor cells. Methods Using multiplex MHC class I tetramers, mass cytometry and mRNA sequencing, we studied EBV‐specific CD8+ T cells at the transcriptomic and phenotypic levels in blood and tumor tissues of the LELC patient. Results Lymphoepithelioma‐like carcinoma lung tumor cells were positive for EBV infection. In both blood and tumor tissues, we detected two populations of EBV‐specific CD8+ T cells targeting the EBV lytic cycle proteins: BRLF1 and BMLF1. Transcriptomic analyses of these two populations in the tumor, which can be considered as tumor‐specific, revealed their distinct exhausted profile and polyclonal TCR repertoire. High‐dimensional phenotypical analysis revealed the distinct phenotype of these cells between blood and tumor tissues. In tumor tissue, EBV‐specific CD8+ TILs were phenotypically heterogeneous, but consistently expressed CD39. Unexpectedly, although the LELC tumor cells expressed abundant PD‐L1, these tumor‐specific CD8+ tumor‐infiltrating lymphocytes (TILs) mostly did not express PD‐1. Conclusion Epstein–Barr virus‐specific CD8+ TILs in EBV‐driven tumor are heterogeneous and partially lack PD‐1 expression, suggesting that anti‐PD1/PD‐L1 immunotherapy may not be an appropriate strategy for disinhibiting EBV‐specific cells in the treatment of LELC patients.
Collapse
Affiliation(s)
- Yannick Simoni
- Vaccine and Infectious Disease Division Fred Hutchinson Cancer Research Center Seattle WA USA.,Agency for Science, Technology and Research Singapore (ASTAR) Singapore Immunology Network (SIgN) Singapore.,ImmunoScape Pte Ltd Singapore
| | - Etienne Becht
- Vaccine and Infectious Disease Division Fred Hutchinson Cancer Research Center Seattle WA USA.,Agency for Science, Technology and Research Singapore (ASTAR) Singapore Immunology Network (SIgN) Singapore
| | - Shamin Li
- Vaccine and Infectious Disease Division Fred Hutchinson Cancer Research Center Seattle WA USA.,Agency for Science, Technology and Research Singapore (ASTAR) Singapore Immunology Network (SIgN) Singapore
| | - Chiew Yee Loh
- Agency for Science, Technology and Research Singapore (ASTAR) Singapore Immunology Network (SIgN) Singapore
| | - Joe Poh Sheng Yeong
- Agency for Science, Technology and Research Singapore (ASTAR) Singapore Immunology Network (SIgN) Singapore.,Department of Anatomical Pathology Singapore General Hospital Singapore General Hospital Singapore
| | - Tony Kiat Hon Lim
- Department of Anatomical Pathology Singapore General Hospital Singapore General Hospital Singapore
| | - Angela Takano
- Department of Anatomical Pathology Singapore General Hospital Singapore General Hospital Singapore
| | - Daniel Shao Weng Tan
- Division of Medical Oncology National Cancer Centre Singapore (NCCS) Singapore.,Agency for Science, Technology and Research (ASTAR) Genome Institute of Singapore (GIS) Singapore
| | - Evan W Newell
- Vaccine and Infectious Disease Division Fred Hutchinson Cancer Research Center Seattle WA USA.,Agency for Science, Technology and Research Singapore (ASTAR) Singapore Immunology Network (SIgN) Singapore.,Senior Corresponding Author
| |
Collapse
|
14
|
Dong XD, Yu J, Feng YY, Ji HY, Yu SS, Liu AJ. Alcohol-soluble polysaccharide from Castanea mollissima blume: Preparation, characteristics and antitumor activity. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103563] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
15
|
Paroder M, Le N, Pham HP, Thibodeaux SR. Important aspects of T‐cell collection by apheresis for manufacturing chimeric antigen receptor T cells. ACTA ACUST UNITED AC 2019. [DOI: 10.1002/acg2.75] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Monika Paroder
- Department of Pathology Montefiore Medical Center of the Albert Einstein College of Medicine Bronx NY USA
| | - Nguyet Le
- Department of Pathology Indiana University School of Medicine Indianapolis IN USA
| | - Huy P. Pham
- Department of Pathology Keck School of Medicine of the University of Southern California Los Angeles CA USA
| | - Suzanne R. Thibodeaux
- Department of Pathology and Immunology Washington University in St. Louis School of Medicine St. Louis MO USA
| |
Collapse
|
16
|
Human Platelet Lysate Media Supplement Supports Lentiviral Transduction and Expansion of Human T Lymphocytes While Maintaining Memory Phenotype. J Immunol Res 2019; 2019:3616120. [PMID: 31565660 PMCID: PMC6746159 DOI: 10.1155/2019/3616120] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 06/14/2019] [Accepted: 07/05/2019] [Indexed: 01/24/2023] Open
Abstract
Immune cell therapy has emerged as a promising approach to treat malignancies that were up until recently only treated on a palliative basis. Chimeric antigen receptor- (CAR-) modified T lymphocytes (T cells) in particular have proven to be very effective for certain hematological malignancies. The production of CAR T cells usually involves viral transduction and ex vivo culture of T cells. The aim of this study was to explore the use of human platelet lysate (HPL) compared to two commonly used supplements, human AB serum (ABS) and fetal bovine serum (FBS), for modified T cell production. For studying transduction, activated T cells were transduced with lentivirus to deliver GFP transgenes with three different promoters. Transduction efficiency (percent GFP) was similar among the supplements, and a modest increase in the transgene product (mean fluorescence intensity) was observed when HPL was used as a supplement compared to ABS. To study the effect of supplements on expansion, peripheral blood mononuclear cells (PBMCs) were activated and expanded in the presence of interleukin 2 (IL2) for fourteen days. T cell expansions using HPL and ABS were comparable and slightly less than the expansion obtained with FBS. Interestingly, cells expanded in media supplemented with HPL showed a higher percentage of T cells with a central memory phenotype compared to those expanded in ABS or FBS. Protein profiling revealed that the phenotypic differences may be explained by elevated levels of several cytokines in HPL, including IL7. The results suggest that the use of HPL as a cell culture supplement during the production of modified T cells is a reasonable alternative to ABS. Furthermore, the use of HPL may enhance in vivo performance of the final product by enriching for central memory T cells that are associated with long-term persistence following adoptive transfer.
Collapse
|
17
|
Coeshott C, Vang B, Jones M, Nankervis B. Large-scale expansion and characterization of CD3 + T-cells in the Quantum ® Cell Expansion System. J Transl Med 2019; 17:258. [PMID: 31391068 PMCID: PMC6686483 DOI: 10.1186/s12967-019-2001-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/23/2019] [Indexed: 12/27/2022] Open
Abstract
Background The rapid evolution of cell-based immunotherapies such as chimeric antigen receptor T-cells for treatment of hematological cancers has precipitated the need for a platform to expand these cells ex vivo in a safe, efficient, and reproducible manner. In the Quantum® Cell Expansion System (Quantum system) we evaluated the expansion of T-cells from healthy donors in a functionally-closed environment that reduces time and resources needed to produce a therapeutic dose. Methods Mononuclear cells from leukapheresis products from 5 healthy donors were activated with anti-CD3/CD28 Dynabeads® and expanded in the Quantum system for 8–9 days using xeno-free, serum-free medium and IL-2. Harvested cells were phenotyped by flow cytometry and evaluated for cytokine secretion by multiplex assays. Results From starting products of 30 or 85 × 106 mononuclear cells, CD3+ T-cell populations expanded over 500-fold following stimulation to provide yields up to 25 × 109 cells within 8 days. T-cell yields from all donors were similar in terms of harvest numbers, viability and doubling times. Functionality (secretion of IFN-γ, IL-2 and TNF-α) was retained in harvested T-cells upon restimulation in vitro and T-cells displayed therapeutically-relevant less-differentiated phenotypes of naïve and central memory T-cells, with low expression of exhaustion markers LAG-3 and PD-1. Conclusions The Quantum system has been successfully used to produce large quantities of functional T-cells at clinical dosing scale and within a short timeframe. This platform could have wide applicability for autologous and allogeneic cellular immunotherapies for the treatment of cancer. Electronic supplementary material The online version of this article (10.1186/s12967-019-2001-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Claire Coeshott
- Terumo BCT, Inc., 10810 West Collins Avenue, Lakewood, CO, 80215, USA.
| | - Boah Vang
- Terumo BCT, Inc., 10810 West Collins Avenue, Lakewood, CO, 80215, USA
| | - Mark Jones
- Terumo BCT, Inc., 10810 West Collins Avenue, Lakewood, CO, 80215, USA
| | - Brian Nankervis
- Terumo BCT, Inc., 10810 West Collins Avenue, Lakewood, CO, 80215, USA
| |
Collapse
|